EP3155003B1 - Constructions d'expression de la protéine bêta-amyloïde et leurs utilisations - Google Patents

Constructions d'expression de la protéine bêta-amyloïde et leurs utilisations Download PDF

Info

Publication number
EP3155003B1
EP3155003B1 EP15806923.7A EP15806923A EP3155003B1 EP 3155003 B1 EP3155003 B1 EP 3155003B1 EP 15806923 A EP15806923 A EP 15806923A EP 3155003 B1 EP3155003 B1 EP 3155003B1
Authority
EP
European Patent Office
Prior art keywords
cell
seq
amyloid beta
gene
expression
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP15806923.7A
Other languages
German (de)
English (en)
Other versions
EP3155003B9 (fr
EP3155003A2 (fr
EP3155003A4 (fr
Inventor
Aftabul Haque
Susan L. Lindquist
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Whitehead Institute for Biomedical Research
Original Assignee
Whitehead Institute for Biomedical Research
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Whitehead Institute for Biomedical Research filed Critical Whitehead Institute for Biomedical Research
Publication of EP3155003A2 publication Critical patent/EP3155003A2/fr
Publication of EP3155003A4 publication Critical patent/EP3155003A4/fr
Publication of EP3155003B1 publication Critical patent/EP3155003B1/fr
Application granted granted Critical
Publication of EP3155003B9 publication Critical patent/EP3155003B9/fr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/80Vectors or expression systems specially adapted for eukaryotic hosts for fungi
    • C12N15/81Vectors or expression systems specially adapted for eukaryotic hosts for fungi for yeasts
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4711Alzheimer's disease; Amyloid plaque core protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5014Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing toxicity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/22Vectors comprising a coding region that has been codon optimised for expression in a respective host
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/001Vector systems having a special element relevant for transcription controllable enhancer/promoter combination
    • C12N2830/002Vector systems having a special element relevant for transcription controllable enhancer/promoter combination inducible enhancer/promoter combination, e.g. hypoxia, iron, transcription factor
    • C12N2830/003Vector systems having a special element relevant for transcription controllable enhancer/promoter combination inducible enhancer/promoter combination, e.g. hypoxia, iron, transcription factor tet inducible
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/008Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • G01N2333/4701Details
    • G01N2333/4709Amyloid plaque core protein
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/2814Dementia; Cognitive disorders
    • G01N2800/2821Alzheimer

Definitions

  • the invention relates to protein chemistry and cellular and molecular biology.
  • Alzheimer's disease is a neurodegenerative disorder characterized by neurofibrillary tangles and plaques containing an amyloid beta peptide. Patients with Alzheimer's disease exhibit progressive dementia and personality dysfunction. Proteolytic cleavage of the amyloid precursor protein (APP) results in the generation of an amyloid beta peptide having a length ranging from 38 to 49 amino acids (e.g., 38-43 amino acids). The amyloid beta 1-42 peptide is particularly prone to self-aggregation and is strongly linked to development of Alzheimer's disease.
  • APP amyloid precursor protein
  • the invention is based, at least in part, on the discovery that a yeast expression construct encoding a polypeptide containing a signal sequence, a Golgi-directing pro sequence, and a human amyloid beta protein can result in significant toxicity when expressed in yeast cells.
  • the invention is also based, at least in part, on the discovery that a mammalian expression construct encoding a polypeptide containing a selected signal sequence and a human amyloid beta protein can result in significant toxicity when expressed in mammalian cells.
  • This discovery permits the carrying out of screening assays using amyloid beta-expressing cells to identify compounds or genetic factors that modulate amyloid beta-induced toxicity. Compounds identified by such screens can be used for the treatment or prevention of neurodegenerative diseases such as Alzheimer's disease.
  • Described herein is an expression construct comprising a promoter operably linked to a nucleic acid encoding a polypeptide comprising a signal sequence, a Golgi-directing pro sequence, and a human amyloid beta protein.
  • an expression construct comprising a promoter operably linked to a nucleic acid encoding a polypeptide comprising a signal sequence, a Golgi-directing pro sequence, and a polypeptide having the formula [X-Y] n , wherein each X is, independently, absent or is a linker; each Y is, independently, a human amyloid beta peptide; and n is an integer between two and eight, inclusive, wherein expression of the nucleic, acid and production of the polypeptide in a yeast cell results in a decrease in growth or viability of the cell.
  • a signal sequence causes the polypeptide containing it to be targeted to the endoplasmic reticulum within a cell.
  • the signal sequence is located at the amino terminus of the polypeptide encoded by the expression construct.
  • the signal sequence can be identical to a naturally occurring signal sequence or can be an artificial (non-naturally occurring) signal sequence.
  • the signal sequence is identical to the signal sequence of a naturally occurring yeast protein.
  • the signal sequence is the yeast mating factor alpha signal sequence.
  • the signal sequence is the signal sequence of yeast killer toxin K1, secreted acid phosphatase, invertase or sucrase, or protoplast secreted protein 1 (Pst1).
  • a Golgi-directing pro sequence causes the transport of the polypeptide containing it to the Golgi.
  • the Golgi-directing pro sequence can be identical to a naturally occurring Golgi-directing pro sequence or can be an artificial (non-naturally occurring) Golgi-directing pro sequence.
  • the Golgi-directing pro sequence is identical to the Golgi-directing pro sequence of a naturally occurring yeast protein.
  • the Golgi-directing pro sequence is the yeast mating factor alpha pro sequence.
  • the Golgi-directing pro sequence is the pro sequence of yeast KEX2, carboxypeptidase Y, Pep4, or Prb1.
  • human amyloid beta protein and "human amyloid beta peptide,” as used interchangeably herein, include proteins whose amino acid sequences are identical to naturally occurring wild type amyloid beta peptides as well as naturally occurring mutant amyloid beta peptides.
  • Wild type amyloid beta peptides include amyloid beta 1-38 (A ⁇ 38), amyloid beta 1-39 (A ⁇ 39), amyloid beta 1-40 (A ⁇ 40), amyloid beta 1-41 (A ⁇ 41), amyloid beta 1-42 (A ⁇ 42), amyloid beta 1-43 (A ⁇ 43), amyloid beta 1-48 (A ⁇ 48), and amyloid beta 1-49 (A ⁇ 49).
  • Amyloid beta mutations include A2T, A2V, H6R, D7N, E11K, F20E, A21G, E22G, E22Q, E22K, E22 deletion, D23N, I31E, E22G/I31E, A42T, and A42V. These mutations may optionally be present in any of the amyloid beta peptides 1-38, 1-39, 1-40, 1-41, 1-42, 1-43, 1-48, and 1-49.
  • a variant of a human amyloid beta protein can be used.
  • a "variant human amyloid beta protein” differs (via substitution, deletion, and/or insertion) from a naturally occurring amyloid beta peptide at up to 10 amino acids (e.g., differs at no more than 5 amino acids, differs at no more than 4 amino acids, differs at no more than 3 amino acids, differs at no more than 2 amino acids, or differs at 1 amino acid) and retains the ability to cause a decrease in growth or viability of a cell when expressed in a fusion polypeptide described herein.
  • an expression construct described herein can optionally be integrated in the genome of the yeast cell.
  • the expression construct can be an integrative plasmid such as pAG303, pAG304, pAG305, pAG306, pRS303, pRS304, pRS305, pRS306, or a derivative thereof.
  • An expression construct described herein can optionally be an episomal plasmid.
  • the promoter can be an inducible promoter such as GAL 1-10, GAL1, GALL, GALS, GPD, ADH, TEF, CYC1, MRP7, MET25, TET, VP16, or VP16-ER.
  • the promoter can be a constitutively active promoter.
  • the polypeptide encoded by an expression construct described herein can include a plurality of human amyloid beta peptides.
  • the polypeptide encoded by an expression construct described herein can optionally contain two, three, four, five, six, seven, eight, or more human amyloid beta peptides.
  • the polypeptide includes at least two human amyloid beta peptides.
  • the polypeptide includes at least four human amyloid beta peptides.
  • each of the human amyloid beta peptides has the same amino acid sequence.
  • the polypeptide includes at least two human amyloid beta peptides having different amino acid sequences.
  • polypeptide encoded by an expression construct described herein can optionally comprise or consist of the amino acid sequence of SEQ ID NO:51, SEQ ID NO:53, SEQ ID NO:55, SEQ ID NO:57, SEQ ID NO:59, SEQ ID NO:61, SEQ ID NO:63, SEQ ID NO:65, SEQ ID NO:67, SEQ ID NO:69, SEQ ID NO:71, SEQ ID NO:73, SEQ ID NO:75, SEQ ID NO:77, SEQ ID NO:79, SEQ ID NO:81, SEQ ID NO:83, or SEQ ID NO:101.
  • yeast cell comprising an expression construct described herein, wherein expression of the nucleic acid and production of the polypeptide in the cell results in a decrease in growth or viability of the cell. In some embodiments, expression of the nucleic acid and production of the polypeptide renders the cell non-viable.
  • a yeast cell can have one or more (e.g., at least two, at least three, or at least four) copies (e.g., integrated or episomal copies) of an expression construct.
  • the yeast is Saccharomyces cerevisiae, Saccharomyces uvae, Saccharomyces kluyveri, Schizosaccharomyces pombe, Kluyveromyces lactis, Hansenula polymorpha, Pichia pastoris, Pichia methanolica, Pichia kluyveri, Yarrowia lipolytica, Candida sp., Candida utilis, Candida cacaoi, Geotrichum sp., or Geotrichum fermentans.
  • At least one gene that encodes a protein involved in drug efflux or cell permeability is disrupted in the yeast cell.
  • the genes PDR1, PDR3, PDR5, SNQ2, or ERG6 can be disrupted in the yeast cell.
  • an expression construct comprising a promoter operably linked to a nucleic acid encoding a polypeptide comprising a signal sequence and a human amyloid beta protein, wherein the signal sequence is an insulin signal sequence (e.g., the human insulin signal sequence) or a trypsin signal sequence (e.g., the human trypsin signal sequence).
  • the human amyloid beta protein is fused directly to the signal sequence.
  • the invention further provides an expression construct comprising a promoter operably linked to a nucleic acid encoding a polypeptide comprising a signal sequence and a polypeptide having the formula [X-Y] n , wherein each X is, independently, absent or is a linker; each Y is, independently, a human amyloid beta peptide; and n is an integer between two and eight, inclusive, wherein the signal sequence is an insulin signal sequence (e.g., the human insulin signal sequence) or a trypsin signal sequence (e.g., the human trypsin signal sequence), wherein expression of the nucleic acid and production of the polypeptide in a yeast cell results in a decrease in growth or viability of the cell.
  • a promoter operably linked to a nucleic acid encoding a polypeptide comprising a signal sequence and a polypeptide having the formula [X-Y] n , wherein each X is, independently, absent or is a linker; each Y is,
  • polypeptide encoded by an expression construct described herein can optionally comprise or consist of the amino acid sequence of SEQ ID NO:93, SEQ ID NO:95, or SEQ ID NO:103.
  • the expression construct is a plasmid or viral vector.
  • a mammalian cell comprising an expression construct comprising a promoter operably linked to a nucleic acid encoding a polypeptide comprising a signal sequence and a human amyloid beta protein, wherein the signal sequence is an insulin signal sequence (e.g., the human insulin signal sequence) or a trypsin signal sequence (e.g., the human trypsin signal sequence), wherein expression of the nucleic acid and production of the polypeptide in the cell results in a decrease in growth or viability of the cell. Expression of the nucleic acid and production of the polypeptide may render the cell non-viable.
  • the cell may be a neuronal cell.
  • the cell may also be a 293T cell.
  • Also disclosed is a method of identifying a genetic suppressor or enhancer of amyloid beta-induced toxicity by: providing a yeast or mammalian cell described herein, wherein an endogenous gene of the cell has been disrupted; culturing the cell under conditions that allow for expression of the protein at a level that, in the absence of disruption of the endogenous gene, is sufficient to induce toxicity in the cell; measuring cell growth or viability in the presence of disruption of the endogenous gene; and comparing cell growth or viability measured in the presence of disruption of the endogenous gene to cell growth or viability in the absence of disruption of the endogenous gene, wherein (i) if cell growth or viability is increased in the presence of disruption of the endogenous gene as compared to in the absence of disruption of the endogenous gene, then the gene is identified as a genetic enhancer of amyloid beta-induced toxicity, and (ii) if cell growth or viability is decreased in the presence of disruption of the endogenous gene as compared to in the absence of disruption of
  • amyloid beta-expressing cells described herein can be used to identify compounds or genetic factors that modulate amyloid beta-induced toxicity.
  • Compounds identified by such screens can be used for the treatment or prevention of neurodegenerative diseases such as Alzheimer's disease.
  • compositions and methods for identifying candidate compounds that prevent or suppress amyloid beta-induced toxicity and genetic suppressors or enhancers of amyloid beta-induced toxicity are compositions and methods for identifying candidate compounds that prevent or suppress amyloid beta-induced toxicity and genetic suppressors or enhancers of amyloid beta-induced toxicity.
  • a yeast expression construct used in the compositions and methods described herein contains a signal sequence, a Golgi-directing pro sequence, and a human amyloid beta protein (i.e., a human amyloid beta peptide).
  • a mammalian expression construct used in the compositions and methods described herein contains a selected signal sequence and a human amyloid beta protein.
  • Described herein is a nucleic acid encoding a polypeptide comprising a signal sequence, a Golgi-directing pro sequence, and a polypeptide having the formula [X-Y] n , wherein each X is, independently, absent or is a linker; each Y is, independently, a human amyloid beta peptide; and n is an integer between two and twenty, inclusive.
  • nucleic acid encoding a polypeptide comprising a signal sequence and a polypeptide having the formula [X-Y] n , wherein each X is, independently, absent or is a linker; each Y is, independently, a human amyloid beta peptide; and n is an integer between two and twenty, inclusive, wherein the signal sequence is an insulin signal sequence (e.g., the human insulin signal sequence) or a trypsin signal sequence (e.g., the human trypsin signal sequence).
  • the signal sequence is an insulin signal sequence (e.g., the human insulin signal sequence) or a trypsin signal sequence (e.g., the human trypsin signal sequence).
  • Human amyloid beta refers to an amino acid sequence identical to human amyloid beta peptide that is derived through proteolytic processing of the human amyloid precursor protein (APP) and is associated with amyloid pathologies.
  • amyloid beta (A ⁇ ) includes naturally occurring wild type amyloid beta peptides as well as naturally occurring mutant amyloid beta peptides.
  • human amyloid beta protein as used herein encompasses proteins produced synthetically by recombinant methods.
  • Wild type amyloid beta peptides include A ⁇ 38, A ⁇ 39, A ⁇ 40, A ⁇ 41, A ⁇ 42, A ⁇ 43, A ⁇ 48, and A ⁇ 49, wherein the number next to A ⁇ denotes the length of the peptide defined by the number of amino acids after proteolytic processing from the amyloid precursor protein.
  • Amyloid beta mutations include A2T, A2V, H6R (English), D7N (Tottori), E1 1K (Leuven), F20E, A21G (Flemish), E22G (Arctic), E22Q (Dutch), E22K (Italian), E22 deletion, D23N (Iowa), I31E, E22G/I3IE, A42T, and A42V.
  • the first letter represents the naturally occurring amino acid
  • the middle number represents the position of the amino acid in the context of the A ⁇ peptide (1-42)
  • the last letter following the numbered position represents the amino acid to which the wild type peptide is mutated.
  • the mutation "E22 deletion” represents an A ⁇ peptide where amino acid glutamic acid (E) at position 22 is deleted and E22G/I31E represents A ⁇ peptide in which both amino acids glutamic acid (E) and isoleucine (I) are replaced by glycine (G) and glutamic acid (E), respectively.
  • These mutations may be present in any of the naturally occurring amyloid beta peptides A ⁇ 38, A ⁇ 39, A ⁇ 40, A ⁇ 41, A ⁇ 42, A ⁇ 43, A ⁇ 48, and A ⁇ 49, or their synthetic derivatives.
  • Amino acids 1-43 of human amyloid beta which amino acids are used as the backbone of the amyloid beta peptides described herein, are as follows: DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGVVIAT (SEQ ID NO:1).
  • amino acids 1-48 of human amyloid beta are used as the backbone of amyloid beta peptides described herein.
  • Amino acids 1-48 of human amyloid beta are as follows: DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGVVIATVIVIT (SEQ ID NO:104).
  • amino acids 1-49 of human amyloid beta are used as the backbone of amyloid beta peptides described herein.
  • Amino acids 1-49 of human amyloid beta are as follows: DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGVVIATVIVITL (SEQ ID NO:105).
  • signal sequence refers to a peptide sequence that is present within a polypeptide and causes the polypeptide to be targeted to the endoplasmic reticulum within a cell.
  • Exemplary signal sequences described in the working examples are the yeast mating factor alpha signal sequence (for a yeast expression construct) and the human insulin and human trypsin signal sequences (for mammalian expression constructs).
  • additional signal sequences that can be used in a yeast expression construct include the signal sequences of killer toxin K1, secreted acid phosphatase, invertase (sucrase), and Pst1.
  • Examples of additional signal sequences that can be used in a mammalian expression construct include the signal sequences of human serum albumin, orexin, human preproparathyroid hormone, somatostatin, shadoo, brain-derived neurotrophic factor, neuropeptide Y, vasoactive intestinal peptide, enkephalin, cholecystokinin, neurotensin, pro-thyrotropin-releasing hormone, neuropeptide W, somatoliberin, and somatotropin.
  • Signal sequences are reviewed in e.g., Wilkinson et al. (1997) J Membr Biol. 155(3):189-97 , Haguenauer-Tsapis (1992) Mol Microbiol. 6(5):573-9 , and Pool (2005) Mol Membr Biol. 22(1-2):3-15 .
  • a polypeptide containing a signal sequence and a human amyloid beta protein may optionally contain one or more heterologous sequences.
  • the heterologous sequence of the fusion protein can optionally be a linker, an immunoglobulin element, a dimerizing domain, a targeting domain, a stabilizing domain, or a purification domain.
  • an amyloid beta protein can be fused with a heterologous sequence such as a detection protein.
  • Exemplary detection proteins include: a fluorescent protein such as green fluorescent protein (GFP), cyan fluorescent protein (CFP) or yellow fluorescent protein (YFP); an enzyme such as ⁇ -galactosidase or alkaline phosphatase (AP); and an epitope such as glutathione-S-transferase (GST) or hemagglutinin (HA).
  • GFP green fluorescent protein
  • CFP cyan fluorescent protein
  • YFP yellow fluorescent protein
  • an enzyme such as ⁇ -galactosidase or alkaline phosphatase (AP)
  • GST glutathione-S-transferase
  • HA hemagglutinin
  • an amyloid beta protein can be fused to GFP at the N- or C-terminus or other parts of the amyloid beta protein.
  • linker refers to a sequence of amino acids linking two polypeptide moieties. Exemplary, non-limiting functions of a linker can include introduction of a flexible component or space-creating region between two protein domains, introduction of proteolytic processing sites (e.g., proteolytic cleavage sites), or creation of an affinity tag.
  • a linker may be any suitable length, for example, a linker may include 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 40, 50, or more amino acids.
  • a linker is a spacer peptide as described herein.
  • the spacer peptide is from the yeast mating factor alpha 1 gene.
  • a linker may include an amino acid sequence selected from SEQ ID NOs:44-49 or a fragment thereof.
  • a polypeptide containing a signal sequence and a human amyloid beta peptide may include a plurality of human amyloid beta peptides.
  • a polypeptide containing a signal sequence, a Golgi-directing pro sequence, and a human amyloid beta peptide may include a plurality of human amyloid beta peptides.
  • the polypeptide encoded by an expression construct described herein can optionally contain two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, twenty, or more human amyloid beta peptides.
  • the polypeptide includes at least two human amyloid beta peptides.
  • the polypeptide includes at least four human amyloid beta peptides. In some embodiments, each of the human amyloid beta peptides includes the same amino acid sequence. For instance, in some embodiments, each of the human amyloid beta peptides is A ⁇ 42. In other embodiments, the polypeptide includes at least two human amyloid beta peptides having different amino acid sequences. For instance, in some embodiments, the polypeptide includes A ⁇ 42 and A ⁇ 43.
  • amyloid beta nucleic acid encompasses a nucleic acid containing a sequence encoding any of the amyloid beta proteins described herein.
  • exemplary amyloid beta nucleic acids include those encoding A ⁇ 42 or A ⁇ 43.
  • nucleic acid generally refers to at least one molecule or strand of DNA, RNA or a derivative or mimic thereof, containing at least one nucleobase, for example, a naturally occurring purine or pyrimidine base found in DNA or RNA.
  • nucleobase for example, a naturally occurring purine or pyrimidine base found in DNA or RNA.
  • nucleic acid refers to at least one single-stranded molecule, but in specific embodiments will also encompass at least one additional strand that is partially, substantially or fully complementary to the at least one single-stranded molecule.
  • a nucleic acid may encompass at least one double-stranded molecule or at least one triple-stranded molecule that comprises one or more complementary strand(s) or "complement(s)" of a particular sequence comprising a strand of the molecule.
  • Yeast strains that can be used in the compositions and methods described herein include, but are not limited to, Saccharomyces cerevisiae, Saccharomyces uvae, Saccharomyces kluyveri, Schizosaccharomyces pombe, Kluyveromyces lactis, Hansenula polymorpha, Pichia pastoris, Pichia methanolica, Pichia kluyveri, Yarrowia lipolytica, Candida sp., Candida utilis, Candida cacaoi, Geotrichum sp., and Geotrichum fermentans.
  • Saccharomyces cerevisiae which ectopically expresses an abnormally processed protein
  • Saccharomyces cerevisiae which ectopically expresses an abnormally processed protein
  • Other yeast strains can be substituted for S. cerevisiae.
  • Certain aspects of the disclosure relate to screening methods for identifying candidate therapeutic agents (e.g., pharmaceutical, chemical, or genetic agents).
  • candidate therapeutic agents e.g., pharmaceutical, chemical, or genetic agents.
  • the methods described herein can optionally be carried out in yeast strains bearing mutations in the ERG6 gene, the PDR1 gene, the PDR3 gene, the PDR5 gene, the SNQ2 gene, and/or any other gene which affects membrane efflux pumps and/or increases permeability for drugs.
  • a nucleic acid encoding a polypeptide described herein may be transfected into a yeast cell using nucleic acid vectors that include, but are not limited to, plasmids, linear nucleic acid molecules, artificial chromosomes, and episomal vectors.
  • Three well known systems used for recombinant plasmid expression and replication in yeast cells include integrative plasmids, low-copy-number ARS-CEN plasmids, and high-copy-number 2 ⁇ plasmids. See Sikorski, "Extrachromosomal cloning vectors of Saccharomyces cerevisiae," in Plasmid, A Practical Approach, Ed. K. G. Hardy, IRL Press, 1993 ; and Yeast Cloning Vectors and Genes, Current Protocols in Molecular Biology, Section II, Unit 13.4, Eds., Ausubel et al., 1994 .
  • integrative plasmids are YIp, which is maintained at one copy per haploid genome, and is inherited in Mendelian fashion.
  • a plasmid containing a gene of interest, a bacterial origin of replication and a selectable gene (typically an antibiotic-resistance marker), is produced in bacteria.
  • the purified vector is linearized within the selectable gene and used to transform competent yeast cells.
  • ARS-CEN plasmids An example of the low-copy-number ARS-CEN plasmids is YCp, which contains the autonomous replicating sequence (ARS1) and a centromeric sequence (CEN4). These plasmids are usually present at 1-2 copies per cell. Removal of the CEN sequence yields a YRp plasmid, which is typically present in 100-200 copies per cell. However, this plasmid is both mitotically and meiotically unstable.
  • ARS1 autonomous replicating sequence
  • CEN4 centromeric sequence
  • 2 ⁇ plasmids An example of the high-copy-number 2 ⁇ plasmids is YEp, which contains a sequence approximately 1 kb in length (named the 2 ⁇ sequence).
  • the 2 ⁇ sequence acts as a yeast replicon giving rise to higher plasmid copy number.
  • these plasmids are unstable and require selection for maintenance. Copy number is increased by having on the plasmid a selection gene operatively linked to a crippled promoter.
  • the plasmid is an integrative plasmid (e.g., pAG303, pAG304, pAG305, pAG306, pRS303, pRS304, pRS305, pRS306, or a derivative thereof).
  • integrative plasmid e.g., pAG303, pAG304, pAG305, pAG306, pRS303, pRS304, pRS305, pRS306, or a derivative thereof.
  • the plasmid is an episomal plasmid (e.g., p426GPD, p416GPD, p426TEF, p423GPD, p425GPD, p424GPD or p426GAL).
  • episomal plasmid e.g., p426GPD, p416GPD, p426TEF, p423GPD, p425GPD, p424GPD or p426GAL.
  • yeast cells are typically transformed by chemical methods (e.g., as described by Rose et al., 1990, Methods in Yeast Genetics, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y .). The cells are typically treated with lithium acetate to achieve transformation efficiencies of approximately 10 4 colony-forming units (transformed cells)/ ⁇ g of DNA. Yeast perform homologous recombination such that the cut, selectable marker recombines with the mutated (usually a point mutation or a small deletion) host gene to restore function. Transformed cells are then isolated on selective media. Of course, any suitable means of introducing nucleic acids into yeast cells can be used.
  • the yeast vectors (plasmids) described herein typically contain a yeast origin of replication, an antibiotic resistance gene, a bacterial origin of replication (for propagation in bacterial cells), multiple cloning sites, and a yeast nutritional gene for maintenance in yeast cells.
  • the nutritional gene (or "auxotrophic marker") is most often one of the following: 1) TRP1 (Phosphoribosylanthranilate isomerase); 2) URA3 (Orotidine-5'-phosphate decarboxylase); 3) LEU2 (3-Isopropylmalate dehydrogenase); 4) HIS3 (Imidazoleglycerolphosphate dehydratase or IGP dehydratase); or 5) LYS2 ( ⁇ -aminoadipate-semialdehyde dehydrogenase).
  • the yeast vectors (plasmids) described herein also contain promoter sequences.
  • a "promoter” is a control sequence that is a region of a nucleic acid sequence at which initiation and rate of transcription are controlled. It may contain genetic elements at which regulatory proteins and molecules may bind, such as RNA polymerase and other transcription factors, to initiate the specific transcription a nucleic acid sequence.
  • the phrases "operatively linked” and “operatively positioned” mean that a promoter is in a correct functional location and/or orientation in relation to a nucleic acid sequence to control transcriptional initiation and/or expression of that sequence.
  • a promoter may be one naturally associated with a nucleic acid sequence, as may be obtained by isolating the 5' non-coding sequences located upstream of the coding segment and/or exon. Such a promoter can be referred to as "endogenous.”
  • a promoter may be a recombinant or heterologous promoter, which refers to a promoter that is not normally associated with a nucleic acid sequence in its natural environment.
  • Such promoters may include promoters of other genes and promoters not "naturally occurring.”
  • the promoters employed may be either constitutive or inducible.
  • yeast-specific promoters may be employed to regulate the expression of a RNA in yeast cells.
  • inducible yeast promoters include GAL1-10, GAL1, GALL, GALS, TET, VP16 and VP16-ER.
  • repressible yeast promoters include Met25.
  • constitutive yeast promoters include glyceraldehyde 3-phosphate dehydrogenase promoter (GPD), alcohol dehydrogenase promoter (ADH), translation-elongation factor-1-alpha promoter (TEF), cytochrome c-oxidase promoter (CYC1), and MRP7.
  • a yeast strain is used that allows for expression, e.g., inducible expression, from GAL promoters on carbon sources other than galactose.
  • the strain carries an integrated or episomal (e.g., plasmid-borne) gene encoding a fusion protein, wherein the Gal4 DNA binding domain is fused to a transcriptional activation domain and a regulatory domain.
  • the fusion protein is characterized in that its ability to activate transcription is regulated by binding of a small molecule to the regulatory domain. For example, in some embodiments, the fusion protein does not activate transcription in the absence of the small molecule, whereas in the presence of the small molecule, the fusion protein activates transcription.
  • Exemplary small molecules include, e.g., steroid hormones, wherein the corresponding regulatory domain comprises at least a portion of a receptor for the small molecule.
  • the small molecule may be an estrogen (e.g., estradiol), or analog thereof (e.g., tamoxifen), and the corresponding regulatory domain comprises at least a portion of the estrogen receptor (ER).
  • Exemplary activation domains include, e.g., viral protein activation domains such as the herpes simplex virus protein VP16 activation domain.
  • the strain carries an integrated or episomal (e.g., plasmid-borne) gene encoding a Gal4-ER-VP16 fusion protein.
  • an estrogen receptor ligand e.g., estradiol
  • GAL promoters on carbon sources other than galactose.
  • Certain aspects of the present disclosure provide methods of screening for a candidate drug (agent or compound) or a genetic factor that modulates amyloid beta-induced toxicity.
  • candidate drugs may be screened by the methods described herein, including nucleic acids, polypeptides, small molecule compounds, and peptidomimetics.
  • genetic agents can be screened by contacting the yeast cell with a nucleic acid construct coding for a gene. For example, one may screen cDNA libraries expressing a variety of genes, to identify genes that modulate amyloid beta-induced toxicity.
  • the identified drugs may modulate amyloid beta-induced toxicity. Accordingly, irrespective of the exact mechanism of action, drugs identified by the screening methods described herein are expected to provide therapeutic benefit to Alzheimer's disease.
  • Certain aspects of the present disclosure provide methods of identifying compounds or genes that modulate amyloid beta-induced toxicity.
  • One of the strongest aspects of yeast is the possibility of performing high throughput screens that may identify genes, peptides and other compounds with the potential to ameliorate toxicity.
  • a large number of compounds can be screened under a variety of growth conditions and in a variety of genetic backgrounds.
  • the toxicity screen has the advantage of not only selecting for compounds that interact with amyloid beta, but also upstream or downstream targets that are not themselves cytotoxic and that are not yet identified.
  • candidate drugs can be screened from large libraries of synthetic or natural compounds.
  • One example is an FDA approved library of compounds that can be used by humans.
  • compound libraries are commercially available from a number of companies including but not limited to Maybridge Chemical Co. (Trevillet, Cornwall, UK), Comgenex (Princeton, NJ), Microsource (New Milford, CT), Aldrich (Milwaukee, WI), AKos Consulting and Solutions GmbH (Basel, Switzerland), Ambinter (Paris, France), Asinex (Moscow, Russia), Aurora (Graz, Austria), BioFocus DPI, Switzerland, Bionet (Camelford, UK), ChemBridge, (San Diego, CA), ChemDiv, (San Diego, CA), Chemical Block Lt, (Moscow, Russia), ChemStar (Moscow, Russia), Exclusive Chemistry, Ltd (Obninsk, Russia), Enamine (Kiev, Ukraine), Evotec (Hamburg, Germany), Indofine (Hillsborough,
  • genomic libraries and disruption libraries can be screened to find extragenic suppressors or enhancers of amyloid beta-induced toxicity. Because the yeast genome is small, 10,000 transformants of each type should be sufficient for good coverage.
  • FRET fluorescent resonance energy transfer
  • an amyloid beta protein can be fused to CFP and to YFP respectively, and integrated in the yeast genome under the regulation of a GAL1-10 promoter.
  • Cells are grown in galactose to induce expression.
  • cells produce the fusion proteins, which aggregate and bring the CFP and YFP close together.
  • the distance between the CFP and YFP is less than the critical value of 100 A that is necessary for FRET to occur.
  • the energy released by the emission of CFP will excite the YFP, which in turn will emit at its characteristic wavelength.
  • FRET based screening can be used to identify candidate compounds including, drugs, genes or other factors that can disrupt the interaction of CFP and YFP by maintaining the proteins in a state that does not allow aggregation to occur.
  • FACS fluorescence activated cell sorting
  • Screens can be carried out under a variety of different conditions.
  • culture media can contain different carbon sources, e.g., different sugars such as glucose, glycerol, galactose, raffinose, etc.
  • multiple screens are performed using two, three, or more different culture conditions (e.g., culture media containing different carbon sources), and compounds or genes identified as "hits" under at least two different culture conditions are identified.
  • screens are performed under two or more different culture conditions (e.g., using culture media containing different carbon sources), wherein the different culture conditions (e.g., different carbon sources) result in different levels of mitochondrial respiration.
  • different culture conditions e.g., different carbon sources
  • growth using culture media containing glucose, glycerol, or galactose result in different levels of mitochondrial respiration.
  • glucose yeast cells ferment and respiration remains low until all glucose is converted to ethanol.
  • galactose respiration is moderately active.
  • glycerol yeast cells are completely dependent on respiration for growth.
  • a screen is performed in parallel using media containing glucose, galactose, or glycerol as a carbon source.
  • Certain embodiments provide methods of further testing those potential drugs that have been identified in the yeast system, in other model systems.
  • the model systems include, but are not limited to, worms, flies, mammalian cells, and in vivo animal models.
  • amyloid beta nucleic acid can be transfected into a mammalian cell using nucleic acid vectors that include, but are not limited to, plasmids, linear nucleic acid molecules, artificial chromosomes, and viral vectors.
  • the vectors can contain as a transgene any of the amyloid beta nucleic acids described herein.
  • the DNA construct(s) can be introduced into an appropriate host cell by any of a variety of suitable means, i.e., transformation, transfection, conjugation, protoplast fusion, electroporation, particle gun technology, calcium phosphate-precipitation, direct microinjection, and the like.
  • the vector is a viral vector (e.g., a lentiviral vector)
  • the vector can be delivered to the cell by direct infection.
  • the vector is stably integrated into the host genome. Methods of producing a cell line containing a stably integrated nucleic acid (e.g., a vector) are well known in the art (see, for example, Sambrook et al.
  • a cell transfected with a nucleic acid can be selected for using a host of antibiotics including, for example, G418, neomycin, or hygromycin B.
  • the transfected nucleic acid contains a suitable antibiotic resistance gene or is co-transfected with a vector containing a suitable antibiotic resistance gene.
  • Amyloid beta can be expressed under the control of an inducible promoter.
  • Methods for assessing the induction of amyloid beta following administration of an inducer include western blotting using an antibody specific for amyloid beta or RT-PCR or northern blotting techniques to detect mRNA expression of amyloid beta. Such methods are well known to those in the art and are described in detail in Sambrook et al. (in Molecular Cloning: A Laboratory Manual. Cold Spring Harbor Laboratory Press, NY, Vol. 1, 2, 3 (1989 )).
  • Expression of amyloid beta can be detected, e.g., at about one hour (e.g., about 30 minutes, about 90 minutes, about two hours, about three hours, about 4 hours, about 6 hours, about 8 hours, about 12 hours, about 12 hours, about 16 hours, about 20 hours, about 24 hours, about 36 hours, about 48 hours or more) post-induction.
  • about one hour e.g., about 30 minutes, about 90 minutes, about two hours, about three hours, about 4 hours, about 6 hours, about 8 hours, about 12 hours, about 12 hours, about 16 hours, about 20 hours, about 24 hours, about 36 hours, about 48 hours or more
  • One can assess (evaluate) a change in expression over time post-induction.
  • the amount of expression of amyloid beta before induction can be compared to the expression of amyloid beta by the cells at various time points (e.g., 1, 4, 8, and 12 hours; 4, 8, 12 and 16 hours; 8, 12, 16, and 20 hours; 12, 24, 36, and 48 hours) post-induction.
  • a suitable starting concentration of an inducing agent is about 0.001 ⁇ M (e.g., about 0.01 ⁇ M, about 0.1 ⁇ M, about 1.0 ⁇ M, about 10.0 ⁇ M, about 100 ⁇ M). It is understood that the concentration of the inducing agent can be optimized for the particular experiment and can depend on, for example, the cell line, the transgene, or the culture conditions the cells are grown in (e.g., low serum conditions).
  • a mammalian cell e.g., a mammalian neuronal cell containing a nucleic acid vector encoding amyloid beta under control of an inducible may be a non-human mammal (e.g., mouse, rat, or guinea pig).
  • the vector can be introduced to the mammalian cell (e.g., a neuronal cell) ex vivo, i.e., the cell can be transfected in vitro and then implanted or otherwise delivered to the mammal (e.g., surgically implanted).
  • a non-human transgenic animal can be established wherein the nucleic acid vector using any of a variety of techniques known in the art (see, for example, Manson et al. (2001) Exp. Rev. Mol. Med. 11 ; and Hofker et al. Transgenic Mouse: Methods and Protocols (Methods in Molecular Biology) Humana Press, Clifton, N.J., Vol. 29 (2002 )).
  • induction of expression of amyloid beta in an animal can be accomplished by administering to the animal an appropriate amount of the inducer.
  • the inducer can be delivered to the mammal as part of food or water (i.e., in the food or water) or can be administered intravenously or parenterally (e.g., subcutaneous injection).
  • Suitable dosages of inducing agent and methods for detecting induction of a transgene in an animal model are described in, for example, Teng et al. (2002) Physiol. Genomics 11:99-107 ; Kim et al. (2003) Am. J. Pathol. 162(5):1693-1707 ; and Zabala et al. (2004) Cancer Res. 64:2799-2804 .
  • a “candidate agent” e.g., a compound or a drug
  • a “candidate agent” is any substance with a potential to reduce, interfere with or curtail (i.e., prevent or suppress) cytotoxicity resulting from overexpression of amyloid beta in a mammalian neuronal cell.
  • candidate agents can be screened by the methods described herein, including, but not limited to, nucleic acids, polypeptides, small molecule compounds, large molecule compounds, peptidomimetics or any other compounds described herein (e.g., see “Compounds” below).
  • the candidate agents are genetic agents that reduce, interfere with, or curtail cytotoxicity resulting from overexpression of amyloid beta in a mammalian neuronal cell.
  • a cDNA library containing coding sequences for a variety of genes can be screened to identify potential therapeutic genes for the diseases described herein.
  • a screen can be performed to identify genetic elements that contribute to cytotoxicity resulting from amyloid beta expression in a mammalian cell.
  • a library of siRNAs or antisense oligonucleotides can be screened such that the level or amount of cytotoxicity in the absence of one or more genes could be determined.
  • a mammalian neuronal cell could be mutagenized to inactivate one or more genes prior to performing the screening assay.
  • a reduced level of amyloid beta-induced cytotoxicity in a cell in the absence of a gene indicates that the gene contributes to amyloid beta-induced cytotoxicity.
  • siRNAs or antisense oligonucleotides that target that gene can be useful in treating Alzheimer's disease.
  • Screening methods to identify an agent capable of preventing or suppressing cytotoxicity resulting from overexpression of amyloid beta can involve the steps of: (i) culturing the cell in the presence of a candidate agent and under conditions that allow for expression of the nucleic acid encoding amyloid beta at a level that, in the absence of the candidate agent, is sufficient to induce toxicity in the cell; (ii) measuring cell viability in the presence of the candidate agent; and (iii) comparing cell viability measured in the presence of the candidate agent to cell viability in the absence of the candidate agent, where if cell viability is greater in the presence of the candidate agent as compared to in the absence of the candidate agent, then the candidate agent is identified as a compound that prevents or suppresses amyloid beta-induced toxicity.
  • the screening assays can involve a mammalian cell containing a stably integrated nucleic acid encoding amyloid beta (optionally under the control of an inducible promoter).
  • the cell can be any mammalian cell, preferably the cell is a neuronal cell (e.g., primary neuronal cells or a neural cell line such as PC12, H4, SK-N-SH, SH-SY5Y, Neuro-2a, SVG p12, CCF-STTG1, SW 1088, SW 1783, LN-18, A172, U-138 MG, T98G, U-87 MG, U-118 MG, Hs 683, M059K, M059J, H4, LN-229, Daoy, or PFSK-1). Additional cell lines are available at the American Type Culture Collection (ATCC), Manassass, VA.
  • ATCC American Type Culture Collection
  • VA Manassass
  • the cells can be treated with two or more concentrations of a compound, where, for example, a concentration-dependence or EC50 is to be determined.
  • Suitable concentrations of a candidate compound for the assay include, for example, about 0.01 ⁇ M to 1 mM of the agent (e.g., about 0.01 ⁇ M to 0.1 ⁇ M, about 0.1 ⁇ M to 1 ⁇ M, about 1 ⁇ M to 10 ⁇ M, about 10 to 1 mM, or about 100 ⁇ M to 1 mM).
  • Methods of assessing the efficacy of an agent to prevent or suppress amyloid beta-induced cytotoxicity can be quantitative, semi-quantitative, or qualitative.
  • the activity of an agent can be determined as a discrete value.
  • An example of a quantitative determination of an agent's is a 50% Effective Concentration, or EC50 value, which is the molar concentration of an agent (e.g., a compound) that gives one-half the maximal response of that agent.
  • EC50 value 50% Effective Concentration, or EC50 value, which is the molar concentration of an agent (e.g., a compound) that gives one-half the maximal response of that agent.
  • the efficacy of an agent can be assessed using a variety of semi-quantitative/qualitative systems known in the art.
  • the efficacy of an agent to prevent or suppress amyloid beta-induced cytotoxicity in a mammalian cell can be expressed as, for example, (a) one or more of “excellent”, “good”, “satisfactory”, “unsatisfactory”, and/or “poor”; (b) one or more of “very high”, “high”, “average”, “low”, and /or “very low”; or (c) one or more of "+++++”, “++++", “+++”, “++", “+”, “+/-", and/or "-".
  • Methods include determining the efficacy of agents in preventing or suppressing amyloid beta-induced cytotoxicity in a mammalian cell (e.g., a compound such as any of those described herein).
  • a mammalian cell e.g., a compound such as any of those described herein.
  • Cells are generally plated on solid support matrix (e.g., a plastic tissue culture plate, or a multi-well (96 or 386-well) tissue culture plate) and grown in appropriate medium. Cells are then contacted with serial dilutions of a candidate agent generally ranging, for example, from 10 ⁇ M to 0.1 ⁇ M concentration.
  • a control compound e.g., a known inhibitor of known concentration
  • a set of cells are grown in the presence of a carrier, buffer, or solvent, in which the compound is delivered.
  • Cells are grown in the presence or absence of test compounds for varying times, for example, from 1 to three days (1 day, 2 days, 3 days, 4 days, 1 week, 2 weeks), followed by a test for the number of cells remaining on the plate or the viability of the cells remaining on the plate.
  • Methods of detecting e.g., determining or measuring) the extent of amyloid beta-induced cytotoxicity in the presence or absence of an agent are myriad and well known to those of ordinary skill in the art. These methods can include, for example, measuring ATP concentration in a cell.
  • the amount of ATP present in a cell or population of cells is proportional to the number of viable cells in that population.
  • ATP concentration can be determined enzymatically, for example, by using luciferase/luciferin. These enzymes produce a light signal in a reaction requiring ATP hydrolysis. Thus, the more ATP present in a sample, the more light produced.
  • cells are first harvested and lysed. Cell lysates are then incubated with luciferase/luciferin and the amount of ATP-dependent light produced from the sample can be detected and/or quantitated using a luminometer (e.g., Turner BioSystems TD-20/20 Luminometer, Turner Biosystems, Sunnyvale, CA).
  • a luminometer e.g., Turner BioSystems TD-20/20 Luminometer, Turner Biosystems, Sunnyvale, CA.
  • the amount of light signal produced from induced cells in the presence of the compound can be compared to the light signal produced from induced cells in the absence of the agent. Where more light signal is produced from lysates of cells cultured in the presence of the agent as compared to cells cultured in the absence of the agent, this indicates that the compound prevents or suppresses cytotoxicity. Further examples of this method are set forth in the Examples.
  • suitable methods for determining the efficacy of agents in preventing or suppressing amyloid beta-induced cytotoxicity include, for example, counting the number of cells remaining after the period of induction in the absence or presence of the agent.
  • cells can be trypsinized from the plate, washed, stained with a dye (e.g., trypan blue), and counted using a microscope or mechanical cell counter (Beckman-Coulter Z1TM Series COULTER COUNTER® Cell and Particle Counter). Since dyes like trypan blue are only taken up by dead or dying cells, this method allows for discrimination (i.e., blue or white cell) between viable and non-viable cells in a population.
  • a dye e.g., trypan blue
  • MTT Diphenyltetrazolium Bromide is a tetrazolium salt (yellowish) that is cleaved to formazan crystals by the succinate dehydrogenase system which belongs to the mitochondrial respiratory chain, and is only active in viable cells.
  • the mitochondrial succinate dehydrogenase reduces the MTT crystals into purple formazan in the presence of an electron coupling reagent.
  • the cells are exposed to the MTT reagent and the more viable cells are present in a well, the more formazan dye is produced.
  • the amount of formazan dye can be measured, for example, using a spectrophotometer.
  • cytotoxicity in a cell e.g., cytotoxicity resulting from overexpression of amyloid beta in a mammalian cell
  • an agent e.g., a compound or a composition described herein
  • induced cells grown in the presence or absence of an agent are also treated with a nucleotide analog that can incorporate into the DNA of the cell upon cell division.
  • nucleotide analogs include, for example, BrdU or 3 H-Thymidine.
  • the amount of label incorporated into the induced cells (grown in the presence and absence of a given agent) is quantitated, and the amount of label incorporation is directly proportional to the amount of cell growth in the population of cells.
  • the amount of label incorporated in the induced cells in the presence and absence of an agent can be normalized to the amount of label incorporated into uninduced cells. More signal (i.e., more DNA synthesis) in an induced cell set treated with the agent as compared to induced cells not treated with the agent indicates that the agent prevents or suppresses amyloid beta-induced cytotoxicity.
  • Suitable methods for assessing suppression or prevention of amyloid beta-induced cytotoxicity by an agent include the detection of apoptosis in a cell.
  • Such methods of detecting or measuring apoptosis include, for example, monitoring DNA fragmentation, caspase activation, or annexin V expression.
  • screening methods described herein can also be used as secondary, or cell-based screens to identify compounds useful in treating a Alzheimer's disease.
  • the screening methods can be used following a primary screen where, for example, a compound is first selected based on an ability to inhibit amyloid beta-induced toxicity in another system (e.g., yeast).
  • Screening assays can be performed in any format that allows for rapid preparation, processing, and analysis of multiple reactions. This can be, for example, in multi-well assay plates (e.g., 96 wells or 386 wells).
  • Stock solutions for various agents can be made manually or robotically, and all subsequent pipetting, diluting, mixing, distribution, washing, incubating, sample readout, data collection and analysis can be done robotically using commercially available analysis software, robotics, and detection instrumentation capable of detecting the signal generated from the assay. Examples of such detectors include, but are not limited to, spectrophotometers, luminometers, fluorimeters, and devices that measure radioisotope decay.
  • Compounds to be screened or identified using any of the methods described herein can include various chemical classes, though typically small organic molecules having a molecular weight in the range of 50 to 2,500 daltons. These compounds can comprise functional groups necessary for structural interaction with proteins (e.g., hydrogen bonding), and typically include at least an amine, carbonyl, hydroxyl, or carboxyl group, and preferably at least two of the functional chemical groups. These compounds often comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures (e.g., purine core) substituted with one or more of the above functional groups.
  • compounds can also include biomolecules including, but not limited to, peptides, polypeptides, peptidomimetics (e.g., peptoids), amino acids, amino acid analogs, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives or structural analogues thereof, polynucleotides, nucleic acid aptamers, and polynucleotide analogs.
  • biomolecules including, but not limited to, peptides, polypeptides, peptidomimetics (e.g., peptoids), amino acids, amino acid analogs, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives or structural analogues thereof, polynucleotides, nucleic acid aptamers, and polynucleotide analogs.
  • Compounds can be identified from a number of potential sources, including: chemical libraries, natural product libraries, and combinatorial libraries comprised of random peptides, oligonucleotides, or organic molecules.
  • Chemical libraries consist of diverse chemical structures, some of which are analogs of known compounds or analogs or compounds that have been identified as “hits” or “leads” in other drug discovery screens, while others are derived from natural products, and still others arise from non-directed synthetic organic chemistry.
  • Natural product libraries are collections of microorganisms, animals, plants, or marine organisms which are used to create mixtures for screening by: (1) fermentation and extraction of broths from soil, plant or marine microorganisms, or (2) extraction of plants or marine organisms.
  • Natural product libraries include polypeptides, non-ribosomal peptides, and variants (non-naturally occurring) thereof.
  • Combinatorial libraries are composed or large numbers of peptides, oligonucleotides, or organic compounds as a mixture. These libraries are relatively easy to prepare by traditional automated synthesis methods, PCR, cloning, or proprietary synthetic methods.
  • non-peptide combinatorial libraries include peptide, protein, peptidomimetic, multiparallel synthetic collection, recombinatorial, and polypeptide libraries.
  • combinatorial chemistry and libraries created therefrom see Myers, Curr. Opin. Biotechnol. 8:701-707 (1997 ).
  • test compounds through the use of the various libraries herein permits subsequent modification of the test compound "hit” or “lead” to optimize the capacity of the "hit” or “lead” to prevent or suppress amyloid beta-induced toxicity and/or amyloid beta-induced aggregation.
  • the compounds identified above can be synthesized by any chemical or biological method.
  • the compounds identified above can also be pure, or may be in a composition (e.g., a pharmaceutical composition) that contains one or more additional component(s), and can be prepared in an assay-, physiologic-, or pharmaceutically- acceptable diluent or carrier (see below).
  • a compound that is found to prevent or suppress amyloid beta-induced toxicity or the formation of amyloid beta aggregates in a cell can be formulated as a pharmaceutical composition, e.g., for administration to a subject to treat a neurodegenerative disease such as Alzheimer's disease.
  • a pharmaceutical composition typically includes a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • the composition can include a pharmaceutically acceptable salt, e.g., an acid addition salt or a base addition salt (see e.g., Berge et al., J. Pharm. Sci. 66:1-19, 1977 ).
  • the compound can be formulated according to standard methods.
  • Pharmaceutical formulation is a well-established art, and is further described, e.g., in Gennaro (ed.), Remington: The Science and Practice of Pharmacy, 20th ed., Lippincott, Williams & Wilkins (2000) (ISBN: 0683306472 ); Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 7th Ed., Lippincott Williams & Wilkins Publishers (1999) (ISBN: 0683305727 ); and Kibbe (ed.), Handbook of Pharmaceutical Excipients American Pharmaceutical Association, 3rd ed. (2000) (ISBN: 091733096X ).
  • a compound that prevents or suppresses amyloid beta-induced toxicity and/or amyloid beta aggregate formation in a cell can be formulated with excipient materials, such as sodium chloride, sodium dibasic phosphate heptahydrate, sodium monobasic phosphate, and a stabilizer. It can be provided, for example, in a buffered solution at a suitable concentration and can be stored at 2-8°C.
  • compositions may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, capsules, pills, powders, liposomes and suppositories.
  • liquid solutions e.g., injectable and infusible solutions
  • dispersions or suspensions tablets, capsules, pills, powders, liposomes and suppositories.
  • the preferred form can depend on the intended mode of administration and therapeutic application.
  • Compositions for the agents described herein may be in the form of injectable or infusible solutions.
  • compositions can be administered by a parenteral mode (e.g., intravenous, subcutaneous, intraperitoneal, or intramuscular injection).
  • parenteral administration e.g., intravenous, subcutaneous, intraperitoneal, or intramuscular injection.
  • parenteral administration e.g., intravenous, subcutaneous, intraperitoneal, or intramuscular injection.
  • parenteral administration e.g., intravenous, subcutaneous, intraperitoneal, or intramuscular injection.
  • the composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable for stable storage at high concentration.
  • Sterile injectable solutions can be prepared by incorporating an agent described herein in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating a compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying that yields a powder of a compound plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin.
  • the compound can be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, and microencapsulated delivery systems.
  • a carrier that will protect the compound against rapid release
  • a controlled release formulation including implants, and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J.R. Robinson, ed., Marcel Dekker, Inc., New York, 1978 .
  • a compound identified as one that prevents or suppresses amyloid beta-induced toxicity and/or amyloid beta aggregate formation in a cell can be modified, e.g., with a moiety that improves its stabilization and/or retention in circulation, e.g., in blood, serum, or other tissues, e.g., by at least 1.5, 2, 5, 10, or 50 fold.
  • the modified compound can be evaluated to assess whether it can reach treatment sites of interest (e.g., locations of aggregate amyloid beta) such as can occur in a cell in a subject with a neurodegenerative disease such as Alzheimer's disease ( e.g., by using a labeled form of the compound).
  • the compound can be associated with a polymer, e.g., a substantially non-antigenic polymer, such as a polyalkylene oxide or a polyethylene oxide. Suitable polymers will vary substantially by weight. Polymers having molecular number average weights ranging from about 200 to about 35,000 Daltons (or about 1,000 to about 15,000, and 2,000 to about 12,500) can be used.
  • a compound can be conjugated to a water soluble polymer, e.g., a hydrophilic polyvinyl polymer, e.g., polyvinylalcohol or polyvinylpyrrolidone.
  • polymers include polyalkylene oxide homopolymers such as polyethylene glycol (PEG) or polypropylene glycols, polyoxyethylenated polyols, copolymers thereof and block copolymers thereof, provided that the water solubility of the block copolymers is maintained.
  • Additional useful polymers include polyoxyalkylenes such as polyoxyethylene, polyoxypropylene, and block copolymers of polyoxyethylene and polyoxypropylene (Pluronics); polymethacrylates; carbomers; and branched or unbranched polysaccharides.
  • the two agents can be formulated separately or together.
  • the respective pharmaceutical compositions can be mixed, e.g., just prior to administration, and administered together or can be administered separately, e.g., at the same or different times.
  • Compounds identified as described herein can be used to treat a subject (e.g., a human subject) that is at risk for or has a disorder associated with amyloid beta mediated toxicity and/or the formation, deposition, accumulation, or persistence of amyloid beta aggregates, e.g., amyloid beta oligomers and/or dimers.
  • a subject e.g., a human subject
  • the disorder is Alzheimer's disease, Down Syndrome, Fragile X syndrome, or systemic amyloidosis.
  • Alzheimer's disease can be diagnosed based on, e.g., patient history (e.g., memory loss), clinical observations, the presence of characteristic neurological and neuropsychological features, and the absence of other conditions that might be responsible for the foregoing.
  • Imaging techniques such as computed tomography (CT), magnetic resonance imaging (MRI), single photon emission computed tomography (SPECT), or positron emission tomography (PET) can be of use. Diagnosis can be confirmed by post-mortem examination of brain material.
  • DSM Diagnostic and Statistical Manual of Mental Disorders
  • NINCDS National Institute of Neurological and Communicative Disorders and Stroke
  • ADRDA Alzheimer's Disease and Related Disorders Association
  • CSF cerebrospinal fluid
  • biomarkers e.g., amyloid beta or tau proteins (e.g., total tau protein and phosphorylated tau) and/or imaging (e.g., PET imaging) with labeled compounds that bind to amyloid beta deposits (e.g., 11C-labeled Pittsburgh Compound-B (11C-PIB) or18F-AV-45 (flobetapir F18)
  • imaging methods may also be of use in the instant invention to assess the in vivo effect of compounds identified herein.
  • a subject has a mutation in a gene encoding amyloid precursor protein (APP), presenilin 1, or presenilin 2.
  • the mutation increases the production of A ⁇ 42 or alters the ratio of A ⁇ 42 to A ⁇ 40.
  • the subject has at least one copy of the ⁇ 4 allele of the apolipoprotein E (APOE) gene.
  • APP amyloid precursor protein
  • presenilin 1 presenilin 2
  • APOE apolipoprotein E
  • Down Syndrome can be diagnosed based on presence of trisomy 21.
  • Fragile X Syndrome is caused by expansion of a trinucleotide gene sequence (CGG) on the X chromosome that results in a failure to express the protein coded by the FMR1 gene, which encodes FMRP. It may be suspected based on the presence of characteristic signs and symptoms, with diagnostic confirmation from genetic testing.
  • CGG trinucleotide gene sequence
  • compositions for treating a subject at risk of (or susceptible to) an amyloid beta mediated disease are described herein.
  • an individual who is at risk of developing Alzheimer's disease and/or has signs suggesting that he or she will develop Alzheimer's disease can be treated with the compounds and methods described herein.
  • treatment is defined as the application or administration of a therapeutic compound to a patient, or application or administration of a therapeutic compound to a subject (e.g., a human subject, who may be referred to as a "patient") who has a disease (or other medically recognized disorder or syndrome), a symptom of disease or a predisposition toward a disease (e.g., one or more risk factors associated with the disease), with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve, or affect (in a manner beneficial to the subject) the disease, the symptoms of disease or the predisposition toward disease.
  • a disease or other medically recognized disorder or syndrome
  • a symptom of disease or a predisposition toward a disease e.g., one or more risk factors associated with the disease
  • treatment is prophylactic, i.e., it is administered to a subject who has not developed the disease (and who may or may not have a predisposition to develop the disease) with an intent to delay, prevent, or reduce the likelihood that the subject will develop the disease or reduce the severity should the subject develop the disease.
  • Compounds may also or alternately be administered to a subject for purposes of testing, research, or diagnosis and/or may be contacted with an isolated tissue, cells, or cell line from a patient, e.g., for purposes of testing, research, diagnosis, or with an intent to subsequently administer the isolated tissue, cells, or cell line to the subject for treatment.
  • Example 1 Amyloid Beta Expression Constructs for Yeast Cells
  • the mating factor alpha 1 gene in yeast encodes a precursor protein of 165 amino acids, called the prepro-alpha-factor.
  • This prepro-alpha-factor consists of a 19-amino acid signal peptide, a 64-amino acid pro region and 4 tandem repeats of alpha-factor sequence, each preceded by a spacer peptide with signature proteolytic cleavage sites.
  • the precursor protein undergoes a series of sequential and defined posttranslational enzymatic modification and proteolytic processing steps in the secretory pathway of the cell to generate the mature 13-amino acid long peptide pheromone, alpha-factor.
  • the nascent prepro-alpha polypeptide is translocated into the lumen of ER, where the signal sequence is cleaved by a signal peptidase to produce the pro-alpha-factor.
  • N-linked carbohydrates are added to three specific glycosylation sites within the pro region, a step that is necessary for the transport of the pro-alpha-factor from the ER to Golgi. Further carbohydrate modification takes place in the Golgi before the final proteolytic processing steps.
  • the first of these three proteolytic processing steps is by the KEX2 endopeptidase that cleaves on the carboxyl side of Lys-Arg (KR) residues at the N terminus of each of the spacer repeats (KREAEA (SEQ ID NO:44) or KREAEAEA (SEQ ID NO:45) or KREADAEA (SEQ ID NO:46)).
  • KREAEA SEQ ID NO:44
  • KREAEAEA SEQ ID NO:45
  • KREADAEA SEQ ID NO:46
  • the human A ⁇ 42 coding sequence was inserted into the mating factor alpha 1 prepro sequence, wherein the 13-amino acid long alpha-factor peptide sequence was replaced by the A ⁇ 42 sequence, leaving all the other sequence components of the prepro-alpha factor intact ( Fig. 1A ).
  • Constructs were prepared based upon the mating factor alpha 1 prepro sequence backbone and containing (i) one (1X), two (2X), four (4X), six (6X), or eight (8X) copies of A ⁇ 42, (ii) four (4X) or six (6X) copies of A ⁇ 40, or four (4X) copies of scrambled A ⁇ 42 ( Fig. 1B ).
  • the nucleotide and amino acid sequences of the fusion polypeptides encoded by the expression constructs are as follows.
  • yeast expression constructs containing four copies (4X) of various A ⁇ 42 mutants were also prepared.
  • the A ⁇ sequence was codon optimized for expression in yeast.
  • the A ⁇ expression construct consists of attB Gateway flanking regions at the 5' and 3' ends (nucleotides ACAAGTTTGTACAAAAAAGCAGGCT (SEQ ID NO:84) and ACCCAGCTTTCTTGTACAAAGTGGT (SEQ ID NO:85) , respectively) for Gateway cloning of MataPrepro-A ⁇ 42 sequence.
  • the entire construct was synthesized into pUC57-Kan plasmid and cloned into the Gateway entry vector pDONR221.
  • Gateway LR cloning Alberti et al., "A suite of Gateway cloning vectors for high throughput genetic analysis in Saccharomyces cerevisiae," Yeast 24:913 (2007 )).
  • the GAL1 promoter enables expression of A ⁇ fusion transgenes under galactose-inducible conditions.
  • the vector has an ampicillin resistant gene for amplification of the vector in E. coli and leucine auxotrophic marker (LEU2) for selection of transformed yeast cells on media lacking leucine ( Fig. 2 ).
  • Yeast cells transformed with either an expression vector containing galactose-inducible green fluorescent protein (GFP) or a galactose-inducible expression plasmid encoding a yeast mating factor alpha signal sequence/human amyloid beta 1-42 (A ⁇ 42) or 1-40 (A ⁇ 40) or scrambled A ⁇ 42 fusion polypeptide were grown on glucose or galactose containing synthetic media and growth was assessed.
  • Transformed cells were selected in medium lacking leucine. Individual transformants were grown overnight in complete synthetic medium (CSM) lacking leucine with 2% raffinose as the carbon source. Cell concentrations (OD600) were adjusted in a 96-well plate to OD 0.5 or 1. Cells were then 5-fold serially diluted and spotted on SD media containing glucose (uninduced) and galactose (induced). Plates were incubated at 30° C for 2 days (glucose) or 3 to 4 days (galactose).
  • GFP galactose-
  • Yeast cells were transformed with galactose-inducible expression vector (pAG425Gal) harboring different mutations in A ⁇ (4 copies). Among these were nine disease-related mutations: English (H6R), Tottori (D7N), Flemish (A21G), Dutch (E22Q), Italian (E22K), Arctic (E22G), E22_deletion, Iowa (D23N), and "wild-type” and two detoxifying mutations: F20E and Arctic_I31E. Growth of transformed yeast cells was tested under induced (galactose) and uninduced (glucose) conditions. Different A ⁇ mutations caused varying degrees of toxicity upon expression under galactose induction ( Fig. 4 ). For example, the Arctic, E22_deletion, and Dutch mutations caused profound toxicity whereas Arctic_I31E and F20E mutations caused significantly lower amount of toxicity ( Fig. 4 ).
  • a human A ⁇ sequence was fused at the C-terminus of the signal sequence of several eukaryotic secretory proteins.
  • the signal sequences were human insulin (Ins), human trypsin (Tryp), human amyloid precursor protein (APP), human placental secreted alkaline phosphatase (SEAP) and Metridia luciferase (mLuc) of the marine copepod Metridia longa.
  • the A ⁇ peptide was fused to these signal peptides without any linker in between the sequences ( Fig. 5 ).
  • human A ⁇ peptide without any secretory signal was also constructed in similar expression vector (labeled "no signal sequence" in Fig. 5 ).
  • nucleotide and amino acid sequences of the fusion polypeptides encoded by the expression constructs are as follows.
  • the human A ⁇ sequence fused to different signal peptide sequence was PCR amplified and cloned in the p3XFLAG-CMV-13 vector.
  • the p3XFLAG-CMV-13 expression vector is a 6.3 kb plasmid for transient expression of fusion proteins in mammalian cells.
  • the prepro-trypsin leader signal sequence which is an integral part of the vector, was deleted by quick change mutagenesis. Deletion of this prepro-trypsin sequence from the 5' of the multiple cloning site enabled use of this vector for cloning of A ⁇ fused to various signal sequences at the N-terminal of the peptide.
  • the vector encodes three adjacent FLAG epitopes downstream of the multiple cloning region.
  • a ⁇ sequence with a stop codon at the C-terminal end was inserted using the EcoR1 and BglII restriction sites so that the Flag tags are not fused to A ⁇ .
  • the human cytomegalovirus (CMV) promoter-regulatory region upstream of the transgene drives transcription of the A ⁇ construct ( Fig. 6 ).
  • the vector also enabled selection of stable transfectants by virtue of the aminoglycoside phosphotransferase II gene (Neo), which confers resistance to aminoglycosides such as G418. It is a shuttle vector for mammalian cells and E. coli, in which the plasmid can be maintained and amplified because of the ampicillin resistant gene (AmpR) in the vector backbone.
  • AmpR ampicillin resistant gene
  • the constructs were transiently transfected in 293T cells using FUGENE® 6 transfection reagent following the manufacture's instruction.
  • 293T cells were routinely propagated in adhesion culture in DMEM medium (Invitrogen) supplemented with 10 % FBS and incubated at 37° C with 5% CO 2 .
  • DMEM medium Invitrogen
  • For transfection of the expression vector cells were plated to approximately 90% confluency (approximately 1.2 ⁇ 10 5 cells/ml) in each well of a 6-well tissue culture plate. Before transfection, the spent medium was replaced by 1 ml of serum free or low serum medium (DMEM without FBS or DMEM + 1% FBS). For each transfection of cells in a single well of a 6-well plate, 1 ⁇ g of the expression construct was used. Upon transfection cells are incubated for up to 96 hours at 37°C with 5% CO 2 .
  • TOXILIGHTTM assay is a bioluminescent, non-destructive cytotoxicity assay designed to measure the release of adenylate kinase from damaged or dying cells.
  • Adenylate kinase is ubiquitous in all eukaryotic cells. It is released into the culture medium when cells are damaged or when cells die. The enzyme actively phosphorylates ADP to form ATP and the resultant ATP is measured using the bioluminescent firefly luciferase reaction.
  • the amount of adenylate kinase in the supernatant is quantitatively proportional to cytolysis and is measured by the intensity of the emitted light by the TOXILIGHTTMreagent.
  • Constructs harboring either A ⁇ 42 or A ⁇ 40 fused to the insulin signal peptide or a vector control without any inserted transgene were used for transfecting the cells.
  • Supernatant from the transfected cells was harvested 48, 72, or 96 hours post-transfection to measure A ⁇ -mediated cytotoxicity.
  • Rat embryonic cortical neurons were used for expressing A ⁇ .
  • an expression vector was used with the human synapsin 1 promoter upstream of the inserted insulin signal peptide-Ap fusion polypeptide transgene ( Fig. 9 ).
  • the encoded fusion polypeptide is processed in the secretory pathway of the neuron, releasing A ⁇ in the extracellular milieu.
  • the pLV-A ⁇ constructs along with the psPAX2 packaging vector and pMD2.G envelope vector were transfected in 293T cells using LIPOFECTAMINE® 2000 (Invitrogen) transfection reagent following the manufacture's instruction to generate lentivirus containing the A ⁇ transgene.
  • the virus was purified using LENTI-XTM Maxi Purification Kit (Clontech) from the supernatant of the transfected 293T cells according to the protocol provided by the manufacturer. Viral titer were determined by using Lentivirus-Associated p24 ELISA Kit (Cell BioLabs).
  • Rat embryonic cortices were dissected out of embryonic (E18) rat brains, and neurons were enriched in adherent monolayer culture. These cortical neurons were infected with lentiviral preparation harboring an A ⁇ transgene or a control gene (RFP in the same vector backbone). Infected cultures were harvested to quantify the amount of A ⁇ in the supernatant and cell lysate by A ⁇ -specific ELISA 21 days after infection ( Fig. 10A ). Corresponding cytotoxicity of the infected neurons was measured by quantifying ATP levels using VIALIGHTTM assay for cell viability (Lonza). Expression of human A ⁇ in the secretory pathway of rat embryonic cortical neurons caused profound cytotoxicity ( Fig. 10B ).
  • rat embryonic cortical neurons were infected with lentiviral constructs (as depicted in Fig. 9 ) expressing wild-type human A ⁇ 42 or various mutations (i.e., F20E, Arctic (E22G), I31E, and E22G/I31E) in the secretory pathway of these cells, differential toxicity was observed, reminiscent of the differential cytotoxic effects caused by these mutations in yeast (see Example 1).
  • the Arctic mutation (E22G) of human A ⁇ 42 had a more toxic effect compared to the wild-type human A ⁇ 42.
  • Three mutations -- F20E, I31E, and E22G/I31E -- exerted less cytotoxic effects compared to either the wild-type human A ⁇ 42 or the Arctic variation of human A ⁇ 42.
  • Rat cortical neurons were infected with a doxycycline-inducible lentiviral GFP construct. Two days post-infection, the cells were infected again with neuron-specific (synapsin promoter) lentiviral A ⁇ constructs (i.e., A ⁇ 40, A ⁇ 42, and various mutations). The doubly-infected cells were treated with doxycycline 7 days post-infection to express GFP. After 21 days of the initial infection, live neurons were visualized using fluorescence microscopy to observe the effect of different A ⁇ variants on neuronal survival and morphology by visualizing neurons expressing GFP in presence of A ⁇ expressed from the transgenes.
  • neuron-specific (synapsin promoter) lentiviral A ⁇ constructs i.e., A ⁇ 40, A ⁇ 42, and various mutations.
  • the doubly-infected cells were treated with doxycycline 7 days post-infection to express GFP. After 21 days of the initial infection, live neurons were visualized using
  • Wild-type human A ⁇ 42 and the A ⁇ 42_Arctic (E22G) mutation caused neuronal death and adversely affected neurite outgrowth and structures, which were detected by microscopic observation of GFP-stained live neurons ( Fig. 11A ).
  • Both the A ⁇ 42_I31E and the A ⁇ 42_Arc_/I31E mutations demonstrated reduced cytotoxicity and no significant morphological distortions of the neurons and neurites in rat cortical neurons infected at multiplicity of infection (MOI) identical to the wild-type A ⁇ 42 and A ⁇ 42_Arctic (E22G) viral constructs.
  • MOI multiplicity of infection
  • wild-type A ⁇ 42 and the A ⁇ 42_Arctic (E22G) mutation caused profound cytotoxicity, resulting in massive neuronal loss and distorted neurite structures and outgrowth ( Fig. 11B ). Also similar to the observations with live neuronal imaging, the A ⁇ 42_I31E and A ⁇ 42_Arc/I31E mutations demonstrated reduced cytotoxicity and had no significant effects on the neuronal morphology ( Figure 11B ).
  • poly-Ap Similar to the poly-Ap (A ⁇ 40 or A ⁇ 42_1X/4X/6X/8X) expression constructs in yeast (see Example 1), a poly-Ap toxicity model was developed in mammalian cells. Stable cell lines with chromosomally-integrated tetracycline (doxycycline)-inducible poly-A ⁇ transgenes (A ⁇ 42-4X, A ⁇ 42-6X, A ⁇ 40-4X, A ⁇ 40-6X and A ⁇ 42-Arctic-4X) linked to an N-terminal insulin signal peptide were generated ( Figs. 13A-13B ). These expression constructs were generated using the pLix402 lentiviral plasmid ( Fig. 12 ).
  • the poly-Ap constructs were individually transfected into 293T cells along with the lentiviral packaging plasmid psPAX and the viral envelope-expressing plasmid mMD2.G using LIPOFECTAMINE® 2000 (Invitrogen) transfection reagent to generate lentivirus containing the poly-Ap transgene.
  • the virus was harvested from the supernatant of transfected 293T cells 2 and 3 days post-transfection and subsequently purified and concentrated using the LENTI-XTM Maxi Purification kit (Clontech) according to the manufacturer's instructions.
  • 293T cells were infected using POLYBRENE® infection reagent (Sigma Aldrich), which is used to introduce retroviral vectors into mammalian cells. Infected cells were then selected using puromycin (2 -10 ⁇ g/ml). Infected cells harboring the transgene survive this selection pressure whereas the control cells without the transgene die in 2-3 days. Aliquots of the selected cells were frozen in liquid nitrogen or maintained and propagated for further applications.
  • POLYBRENE® infection reagent Sigma Aldrich
  • a ⁇ peptide from these stable cell lines was measured in the culture supernatant 72-96 hours following doxycycline induction ( Fig. 13D ). Toxicity was measured 72-96 hours following doxycycline induction using a TOXILIGHTTM cytotoxicity assay (Lonza) as described in Example 2.
  • TOXILIGHTTM cytotoxicity assay Lionza
  • These stable lines demonstrated an A ⁇ peptide-mediated cytotoxic effect (A ⁇ 42-6X ⁇ A ⁇ 42-Arctic-4X > A ⁇ 42-4X > AB40-6X ⁇ A ⁇ 40-4X), as determined by adenylate kinase release in the cell culture supernatant from damaged cells upon doxycycline-induction of the transgenes ( Fig. 13C ).
  • Rat embryonic cortical neurons were infected with lentiviral poly-Ap expression constructs to model poly-Ap toxicity.
  • Tetracycline (doxycycline)-inducible poly-Ap (A ⁇ 42-4X, A ⁇ 42-6X, A ⁇ 40-4X, A ⁇ 40-6X, and A ⁇ 42-Arctic-4X) viral constructs described in Example 5 were used to express poly-Ap constructs in rat cortical neurons.
  • the poly-Ap coding sequence is linked to an N-terminal insulin signal peptide.
  • the poly-Ap expression plasmids were packaged in lentiviral particles, which were generated by 293T cells as described in Example 3.
  • Rat cortical neurons were enriched from cortices dissected out of embryonic (E18) rat brains and enriched in adherent monolayer culture as described in Example 3. Purified lentiviral preparations at different MOI were used to infect these cortical neurons to express poly-Ap transgenes. Three days post-infection, expression of poly-A ⁇ was induced by adding doxycycline (2 ⁇ g/ml) to the infected cultures. The viability of the infected neurons was measured three weeks post doxycycline-induction by quantifying ATP levels using VIALIGHTTM assay for cell viability (Lonza) to determine the cytotoxicity of the poly-Ap expression constructs. Expression of human A ⁇ 42-4X-WT or A ⁇ 42-4X-Arctic in the secretory pathway of rat embryonic cortical neurons caused profound cytotoxicity compared to A ⁇ 40-4X ( Fig. 14 ).
  • the cytotoxic model in 293T cells was used to test the effect of small molecule compounds to alleviate A ⁇ -mediated toxicity.
  • the A ⁇ 42 or A ⁇ 40 expression construct was transiently transfected and small molecules were added in different dosages 12 hours post-transfection. Cytotoxicity was measured at different compound dosages by adenylate kinase released from the damaged cells by TOXILIGHTTM BioAssay Kit (Lonza) 96 hours post-transfection.
  • One example of a compound-mediated effect of toxicity alleviation was by clioquinol (CQ), which rescued A ⁇ -mediated toxicity between concentrations of 2 and 5 ⁇ M ( Fig. 15 ).
  • CQ clioquinol
  • Similar assays are also performed using virally-infected stable 293T cells harboring tetracycline (doxycycline)-inducible poly-Ap expression constructs to identify compounds that rescue cytotoxicity caused by poly-Ap expression constructs.
  • Example 8 An A ⁇ 43 Expression Construct is Toxic to Yeast Cells
  • a ⁇ 43 was expressed in the yeast S. cerevisiae as a tandem polypeptide (four tandem A ⁇ peptides linked by 6-8 amino acid linkers) using a poly-Ap expression construct under the control of the galactose-inducible GAL1 promoter.
  • the A ⁇ polypeptide coding sequence is linked at the N-terminus to the mating factor ⁇ pre-pro signal sequence for directed expression of the transgene in the secretory pathway ( Fig. 16A ).
  • the yeast expression construct containing four copies (4X) of A ⁇ 43 had the following nucleotide and amino acid sequences.
  • the toxicity of A ⁇ 43 to yeast cells was compared to that of similar poly-Ap expression constructs expressing A ⁇ 42, A ⁇ 40, A ⁇ 48, or A ⁇ 49.
  • the A ⁇ _scrambled construct in which the amino acid sequence of A ⁇ 42 was randomly scrambled, served as a control tandem peptide, and a GFP construct served as a vector control.
  • the transgenes were expressed from an episomal expression plasmid (2-micron) under the selection of leucine auxotrophy.
  • the growth of the transformed yeast was measured on synthetic agar media lacking leucine and containing galactose for induction of the transgene.
  • the transformed strains were also grown on similar media with glucose as a measure of growth without the induction of transgene expression.
  • An ImageJ spot assay quantification macro was used to quantify yeast growth on solid media.
  • a ⁇ 43 was significantly more cytotoxic than other A ⁇ peptides when expressed in yeast ( Fig. 16B ).
  • the relative toxicity was A ⁇ 43 > A ⁇ 49 ⁇ A ⁇ 48 ⁇ A ⁇ 42 > A ⁇ 40.
  • Example 9 An A ⁇ 43 Expression Construct is Toxic to Mammalian Cells
  • an expression construct was generated by fusing the insulin signal peptide to the N-terminus of the A ⁇ 43 peptide coding sequence ( Figs. 17A and 17B ).
  • the mammalian expression construct encoding A ⁇ 43 had the following nucleotide and amino acid sequences.
  • the expression construct is expressed under the CMV promoter using pCMV vector as described above (see Example 2 and Fig. 6 ).
  • the cytotoxic effect of A ⁇ 43 in mammalian cells was tested in 293T cells by transiently transfecting the expression construct and measuring adenylate kinase released from the damaged cells 96 hours post-transfection. Cytotoxicity was measured using the TOXILIGHTTM BioAssay Kit (Lonza) following the manufacturer's instructions (see Example 2). Similar to the results from yeast cells described in Example 8, A ⁇ 43 was significantly more cytotoxic compared to A ⁇ 42, A ⁇ 48, A ⁇ 49, or A ⁇ 40 ( Fig. 17C ).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Toxicology (AREA)
  • Immunology (AREA)
  • Plant Pathology (AREA)
  • Mycology (AREA)
  • Medicinal Chemistry (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Analytical Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Neurology (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Cell Biology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Claims (10)

  1. Construction d'expression comprenant un promoteur lié de manière fonctionnelle à un acide nucléique codant pour un polypeptide comprenant une séquence de signal, une séquence pro d'orientation de Golgi, et un polypeptide ayant la formule [X-Y]n, dans laquelle chaque X est, indépendamment, absent ou est un lieur ; chaque Y est, indépendamment, un peptide bêta-amyloïde humain ; et n est un nombre entier entre deux et huit, inclus, dans laquelle une expression de l'acide nucléique et une production du polypeptide dans une cellule de levure résultent en une diminution de croissance ou de viabilité de la cellule.
  2. Construction d'expression selon la revendication 1, dans laquelle :
    (i) la séquence de signal est identique à la séquence de signal d'une protéine de levure présente naturellement, facultativement dans laquelle la séquence de signal est la séquence de signal alpha de facteur de conjugaison de levure ou la séquence de signal de toxine tueuse de levure K1, de phosphatase acide sécrétée, d'invertase (sucrase), ou de Pst1 ;
    (ii) la séquence pro d'orientation de Golgi est la séquence pro alpha de facteur de conjugaison de levure ou la séquence pro de KEX2 de levure, de carboxypeptidase Y, de Pep4, ou de Prb1 ;
    (iii) le promoteur est un promoteur inductible, facultativement dans laquelle le promoteur inductible est GAL1-10, GAL1, GALL, GALS, GPD, ADH, TEF, CYC1, MRP7, MET25, TET, VP16, ou VP16-ER ; et/ou
    (iv)
    a) chaque protéine bêta-amyloïde humaine est indépendamment choisie dans le groupe constitué d'Aβ38, de type sauvage, Aβ39 de type sauvage, Aβ40de type sauvage, Aβ41 de type sauvage, Aβ42de type sauvage, Aβ43de type sauvage, Aβ48 de type sauvage, et Aβ49 de type sauvage ;
    b) chaque protéine bêta-amyloïde humaine est indépendamment choisie dans le groupe constitué d'Aβ38, Aβ39, Aβ40, Aβ41, Aβ42, Aβ43, Aβ48 et Aβ49 et comprend une mutation choisie dans le groupe constitué d'A2T, A2V, H6R, D7N, E11K, F20E, A21G, E22G, E22Q, E22K, délétion de E22, D23N, I31E, E22G/I31E, A42T et A42V, dans laquelle la mutation est par rapport à la séquence d'acides aminés de SEQ ID NO:105 ; ou
    c) la protéine bêta-amyloïde humaine est Aβ42 de type sauvage.
  3. Construction d'expression selon la revendication 1 ou 2, dans laquelle le polypeptide comprend :
    (i) au moins quatre peptides bêta-amyloïdes humains, et/ou
    (ii)
    a) chaque peptide bêta-amyloïde humain comprend la même séquence d'acides aminés ; ou
    b) le polypeptide comprend au moins deux peptides bêta-amyloïdes humains ayant des séquences d'acides aminés différentes.
  4. Construction d'expression selon l'une quelconque des revendications 1 à 3, dans laquelle :
    (i) le polypeptide comprend la séquence d'acides aminés de SEQ ID NO:53, SEQ ID NO:55, SEQ ID NO:57, SEQ ID NO:59, SEQ ID NO:61, SEQ ID NO:63, SEQ ID NO:65, SEQ ID NO:67, SEQ ID NO:69, SEQ ID NO:71, SEQ ID NO:73, SEQ ID NO:75, SEQ ID NO:77, SEQ ID NO:79, SEQ ID NO:81, SEQ ID NO:83, ou SEQ ID NO:101 ; et/ou
    (ii) la construction d'expression est un plasmide épisomique ou un plasmide intégratif, facultativement dans laquelle le plasmide intégratif est pAG303, pAG304, pAG305, pAG306, pRS303, pRS304, pRS305, pRS306, ou un dérivé de ceux-ci.
  5. Cellule de levure comprenant la construction d'expression selon l'une quelconque des revendications précédentes.
  6. Cellule de levure selon la revendication 6, dans laquelle :
    (i) une expression de l'acide nucléique et une production du polypeptide rendent la cellule non viable ;
    (ii) la construction d'expression est épisomique ou est intégrée dans le génome de la cellule de levure ;
    (iii) la levure est Saccharomyces cerevisiae, Saccharomyces uvae, Saccharomyces kluyveri, Schizosaccharomyces pombe, Kluyveromyces lactis, Hansenula polymorpha, Pichia pastoris, Pichia methanolica, Pichia kluyveri, Yarrowia lipolytica, Candida sp., Candida utilis, Candida cacaoi, Geotrichum sp., ou Geotrichum fermentans ;
    (iv) au moins un gène qui code pour une protéine impliquée dans un efflux de médicament ou une perméabilité de cellule est perturbé, facultativement dans laquelle l'au moins un gène est PDR1, PDR3, PDR5, SNQ2, ou ERG6 ; et/ou
    (v) la cellule de levure permet l'expression à partir de promoteurs GAL sur des sources de carbone autres que galactose ; facultativement dans laquelle la cellule de levure comprend un gène codant pour une protéine hybride comprenant le domaine de liaison d'ADN Gal4 fusionné à un domaine d'activation transcriptionnelle et un domaine régulateur ; facultativement dans laquelle la protéine hybride active une transcription en présence d'oestrogène ou d'un analogue de celui-ci ; de préférence dans laquelle la cellule de levure comprend un gène codant pour une protéine de fusion Gal4-ER-VP16.
  7. Procédé d'induction de toxicité dans une cellule, le procédé comprenant :
    la fourniture de la cellule selon la revendication 5 ou 6 ; et
    l'induction d'un niveau d'expression de l'acide nucléique dans la cellule qui est toxique pour la cellule.
  8. Procédé d'identification d'un composé qui empêche ou supprime une toxicité induite par bêta-amyloïdes, le procédé comprenant :
    la culture de la cellule selon la revendication 5 ou 6, en présence d'un agent candidat et dans des conditions qui permettent l'expression de l'acide nucléique à un niveau qui, en l'absence de l'agent candidat, est suffisant pour induire une toxicité dans la cellule ;
    la mesure de croissance ou viabilité cellulaire en présence de l'agent candidat ; et
    la comparaison de la croissance ou viabilité cellulaire mesurée en présence de l'agent candidat à une croissance ou viabilité cellulaire en l'absence de l'agent candidat,
    dans lequel si la croissance ou viabilité cellulaire est accrue en présence de l'agent candidat par rapport à en l'absence de l'agent candidat, alors l'agent candidat est identifié en tant que composé qui empêche ou supprime une toxicité induite par bêta-amyloïdes.
  9. Procédé d'identification d'un suppresseur ou amplificateur génétique de toxicité induite par bêta-amyloïdes, le procédé comprenant :
    la fourniture de la cellule selon la revendication 5 ou 6, dans lequel la cellule a été génétiquement modifiée pour surexprimer un gène ;
    la culture de la cellule dans des conditions qui permettent l'expression de la protéine à un niveau qui, en l'absence de surexpression du gène, est suffisant pour induire une toxicité dans la cellule ;
    la mesure de la croissance ou viabilité cellulaire en présence de surexpression du gène ; et
    la comparaison de la croissance ou viabilité cellulaire mesurée en présence de surexpression du gène à la croissance ou viabilité cellulaire en l'absence de surexpression du gène,
    dans lequel (i) si la croissance ou viabilité cellulaire est accrue en présence d'une surexpression du gène par rapport à en l'absence de surexpression du gène, alors le gène est identifié en tant que suppresseur génétique de toxicité induite par bêta-amyloïdes, et (ii) si la croissance ou viabilité cellulaire est diminuée en présence de surexpression du gène par rapport à en l'absence de surexpression du gène, alors le gène est identifié en tant qu'amplificateur génétique de toxicité induite par bêta-amyloïdes.
  10. Procédé d'identification d'un suppresseur ou amplificateur génétique de toxicité induite par bêta-amyloïdes, le procédé comprenant :
    la fourniture de la cellule selon la revendication 5 ou 6, dans lequel un gène endogène de la cellule a été perturbé ;
    la culture de la cellule dans des conditions qui permettent l'expression de la protéine à un niveau qui, en l'absence de perturbation du gène endogène, est suffisant pour induire une toxicité dans la cellule ;
    la mesure de la croissance ou viabilité cellulaire en présence de perturbation du gène endogène ; et
    la comparaison de la croissance ou viabilité cellulaire mesurée en présence de perturbation du gène endogène à la croissance ou viabilité cellulaire en l'absence de perturbation du gène endogène,
    dans lequel (i) si la croissance ou viabilité cellulaire est accrue en présence de perturbation du gène endogène par rapport à en l'absence de perturbation du gène endogène, alors le gène est identifié en tant qu'amplificateur génétique de toxicité induite par bêta-amyloïdes, et (ii) si la croissance ou viabilité cellulaire est diminuée en présence de perturbation du gène endogène par rapport à en l'absence de perturbation du gène endogène, alors le gène est identifié en tant que suppresseur génétique de toxicité induite par bêta-amyloïdes.
EP15806923.7A 2014-06-13 2015-06-12 Constructions d'expression de la protéine bêta-amyloïde et leurs utilisations Active EP3155003B9 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462012107P 2014-06-13 2014-06-13
PCT/US2015/035571 WO2015192009A2 (fr) 2014-06-13 2015-06-12 Constructions d'expression de la protéine bêta-amyloïde et leurs utilisations

Publications (4)

Publication Number Publication Date
EP3155003A2 EP3155003A2 (fr) 2017-04-19
EP3155003A4 EP3155003A4 (fr) 2018-04-18
EP3155003B1 true EP3155003B1 (fr) 2020-08-05
EP3155003B9 EP3155003B9 (fr) 2020-12-30

Family

ID=54834571

Family Applications (1)

Application Number Title Priority Date Filing Date
EP15806923.7A Active EP3155003B9 (fr) 2014-06-13 2015-06-12 Constructions d'expression de la protéine bêta-amyloïde et leurs utilisations

Country Status (5)

Country Link
US (1) US9822156B2 (fr)
EP (1) EP3155003B9 (fr)
JP (1) JP6655557B2 (fr)
KR (1) KR102342622B1 (fr)
WO (1) WO2015192009A2 (fr)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3191578B1 (fr) 2014-09-12 2020-11-04 Whitehead Institute for Biomedical Research Cellules exprimant l'apolipoprotéine e et leurs utilisations
CN107530451A (zh) * 2015-05-07 2018-01-02 南佛罗里达大学 用于天使人综合征的基因治疗方法的修饰的ube3a基因
KR20200107545A (ko) 2019-03-08 2020-09-16 정하음 건축물의 문 시공방법
EP4286405A1 (fr) * 2021-01-28 2023-12-06 Abrain Thérapie génique pour le traitement de maladies neurodégénératives

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE69940785D1 (de) * 1998-11-20 2009-06-04 Fuso Pharmaceutical Ind Proteinexpressionsvektor und benutzung desselbigen
EP1187920A2 (fr) 1999-06-09 2002-03-20 Arch Development Corporation Genes, proteines et matieres de recombinaison de type prion et procedes associes
MXPA03004853A (es) 2000-12-05 2005-02-14 Penn State Res Found Metodos y composiciones para produccion sumamente eficiente de proteinas heterologas en levadura.
JP4344138B2 (ja) 2001-02-15 2009-10-14 ユニバーシティ オブ シカゴ タンパク質の折り畳みに影響する薬剤の酵母スクリーニング
JP4056248B2 (ja) * 2001-11-30 2008-03-05 武田薬品工業株式会社 アミロイドβタンパク質産生阻害薬のスクリーニング方法
WO2005031363A1 (fr) * 2003-09-24 2005-04-07 University Of Chicago Preparation membranaire tiree de pichia pastoris pour tester l'activite gamma-secretase
WO2007064917A2 (fr) 2005-11-30 2007-06-07 Abbott Laboratories Procedes de preparation de formes recombinees de la proteine beta-amyloide humaine et utilisation desdites proteines
GB0700645D0 (en) 2007-01-12 2007-02-21 Eisai London Res Lab Ltd Targets for disease therapy
CN101492674A (zh) 2008-01-21 2009-07-29 吉林圣元科技有限责任公司 用甲基营养酵母生产人淀粉样蛋白Aβ1-42的方法
US9677079B2 (en) * 2010-01-12 2017-06-13 Whitehead Insititute for Biomedical Research Yeast cells expressing amyloid beta and uses therefor
US20130045483A1 (en) * 2011-07-01 2013-02-21 Whitehead Institute For Biomedical Research Yeast cells expressing amyloid beta and uses therefor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None *

Also Published As

Publication number Publication date
KR20170012547A (ko) 2017-02-02
JP6655557B2 (ja) 2020-02-26
JP2017517269A (ja) 2017-06-29
EP3155003B9 (fr) 2020-12-30
KR102342622B1 (ko) 2021-12-22
EP3155003A2 (fr) 2017-04-19
EP3155003A4 (fr) 2018-04-18
US20150361148A1 (en) 2015-12-17
WO2015192009A2 (fr) 2015-12-17
US9822156B2 (en) 2017-11-21
WO2015192009A3 (fr) 2016-03-17

Similar Documents

Publication Publication Date Title
US10240160B2 (en) Yeast cells expressing amyloid beta and uses therefor
US10428335B2 (en) Yeast cells expressing TAR DNA-binding protein 43 and uses therefor
Sekito et al. RTG-dependent mitochondria-to-nucleus signaling is regulated by MKS1 and is linked to formation of yeast prion [URE3]
US20130045483A1 (en) Yeast cells expressing amyloid beta and uses therefor
EP3155003B1 (fr) Constructions d'expression de la protéine bêta-amyloïde et leurs utilisations
US20120003243A1 (en) Yeast ectopically expressing abnormally processed proteins and uses therefor
US9128081B2 (en) TDP-43-storing cell model
JP2009526541A (ja) アポトーシス方法、遺伝子およびタンパク質
US20130237451A1 (en) Compositions and methods for treatment of protein misfolding diseases
US20100196932A1 (en) Yeast Reporter System
US8192986B2 (en) Compositions and methods for treatment of protein misfolding diseases
EP2162541B1 (fr) Procédé de criblage pour des composés réducteurs de stress er
JP2017517269A5 (fr)
JP2010506569A (ja) α−シヌクレインを発現する細胞およびその使用
WO2015066776A2 (fr) Modèle de levure pour une toxicité synergique

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20161207

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: WHITEHEAD INSTITUTE FOR BIOMEDICAL RESEARCH

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
RIC1 Information provided on ipc code assigned before grant

Ipc: G01N 33/50 20060101ALI20171128BHEP

Ipc: C12N 15/81 20060101ALI20171128BHEP

Ipc: C07K 14/47 20060101AFI20171128BHEP

Ipc: C12N 15/85 20060101ALI20171128BHEP

RIC1 Information provided on ipc code assigned before grant

Ipc: C12N 15/85 20060101ALI20171208BHEP

Ipc: C12N 15/81 20060101ALI20171208BHEP

Ipc: G01N 33/50 20060101ALI20171208BHEP

Ipc: C07K 14/47 20060101AFI20171208BHEP

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1235797

Country of ref document: HK

A4 Supplementary search report drawn up and despatched

Effective date: 20180321

RIC1 Information provided on ipc code assigned before grant

Ipc: C12N 15/81 20060101ALI20180315BHEP

Ipc: C07K 14/47 20060101AFI20180315BHEP

Ipc: G01N 33/50 20060101ALI20180315BHEP

Ipc: C12N 15/85 20060101ALI20180315BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20190409

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20200430

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1298598

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200815

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602015057044

Country of ref document: DE

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: VALIPAT S.A. C/O BOVARD SA NEUCHATEL, CH

REG Reference to a national code

Ref country code: CH

Ref legal event code: PK

Free format text: BERICHTIGUNG B9

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20200805

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1298598

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200805

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201207

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201105

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201106

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201105

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201205

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602015057044

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

26N No opposition filed

Effective date: 20210507

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210612

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210612

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20150612

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20230626

Year of fee payment: 9

Ref country code: DE

Payment date: 20230626

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: BE

Payment date: 20230627

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20230627

Year of fee payment: 9

Ref country code: CH

Payment date: 20230702

Year of fee payment: 9

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200805